1
|
Comino Garcia-Munoz A, Varlet I, Grau GE, Perles-Barbacaru TA, Viola A. Contribution of Magnetic Resonance Imaging Studies to the Understanding of Cerebral Malaria Pathogenesis. Pathogens 2024; 13:1042. [PMID: 39770302 PMCID: PMC11728472 DOI: 10.3390/pathogens13121042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 01/16/2025] Open
Abstract
Cerebral malaria (CM), the most lethal clinical syndrome of Plasmodium falciparum infection, mostly affects children under 5 in sub-Saharan Africa. CM is characterized by seizures and impaired consciousness that lead to death in 15-20% of cases if treated quickly, but it is completely fatal when untreated. Brain magnetic resonance imaging (MRI) is an invaluable source of information on the pathophysiology of brain damage, but, due to limited access to scanners in endemic regions, only until very recently have case reports of CM patients studied with advanced MRI methods been published. The murine model of experimental cerebral malaria (ECM) shares many common features with the human disease and has been extensively used to study the pathogenic mechanisms of the neurological syndrome. In vivo MRI studies on this model, the first of which was published in 2005, have contributed to a better understanding of brain lesion formation in CM and identified disease markers that were confirmed by MRI studies published from 2013 onwards in pediatric patients from endemic areas. In this review, we recapitulate the main findings and critically discuss the contributions of MRI studies in the ECM model to the understanding of human CM.
Collapse
Affiliation(s)
- Alicia Comino Garcia-Munoz
- Centre de Résonance Magnétique Biologique et Médicale (CRMBM) UMR 7339, Faculté des Sciences Médicales et Paramédicales la Timone, Aix-Marseille Université, CNRS, 13055 Marseille, France; (A.C.G.-M.); (I.V.); (T.-A.P.-B.)
| | - Isabelle Varlet
- Centre de Résonance Magnétique Biologique et Médicale (CRMBM) UMR 7339, Faculté des Sciences Médicales et Paramédicales la Timone, Aix-Marseille Université, CNRS, 13055 Marseille, France; (A.C.G.-M.); (I.V.); (T.-A.P.-B.)
| | - Georges Emile Grau
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine, The University of Sydney, Medical Foundation Building (K25), Camperdown, NSW 2042, Australia;
| | - Teodora-Adriana Perles-Barbacaru
- Centre de Résonance Magnétique Biologique et Médicale (CRMBM) UMR 7339, Faculté des Sciences Médicales et Paramédicales la Timone, Aix-Marseille Université, CNRS, 13055 Marseille, France; (A.C.G.-M.); (I.V.); (T.-A.P.-B.)
| | - Angèle Viola
- Centre de Résonance Magnétique Biologique et Médicale (CRMBM) UMR 7339, Faculté des Sciences Médicales et Paramédicales la Timone, Aix-Marseille Université, CNRS, 13055 Marseille, France; (A.C.G.-M.); (I.V.); (T.-A.P.-B.)
| |
Collapse
|
2
|
Wang J, Zhu Q, Shen Y, Liang J, Wang Y, Huang Y, Tong G, Wang X, Zhang N, Yu K, Li Y, Zhao Y. CD8 + T cell infiltration and proliferation in the brainstem during experimental cerebral malaria. CNS Neurosci Ther 2024; 30:e14431. [PMID: 37697956 PMCID: PMC10916431 DOI: 10.1111/cns.14431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/06/2023] [Accepted: 07/16/2023] [Indexed: 09/13/2023] Open
Abstract
INTRODUCTION Cerebral malaria (CM) is a lethal neuroinflammatory disease caused by Plasmodium infection. Immune cells and brain parenchyma cells contribute to the pathogenesis of CM. However, a systematic examination of the changes that occur in the brain parenchyma region during CM at the single-cell resolution is still poorly studied. AIMS To explore cell composition and CD8+ T cell infiltration, single-cell RNA sequencing (scRNA-seq) was performed on the brainstems of healthy and experimental cerebral malaria (ECM) mice. Then CD8+ T cell infiltration was confirmed by flow cytometry and immunofluorescence assays. Subsequently, the characteristics of the brain-infiltrated CD8+ T cells were analyzed. Finally, the interactions between parenchyma cells and brain-infiltrated CD8+ T cells were studied with an astrocytes-CD8+ T cell cocultured model. RESULTS The brainstem is the most severely damaged site during ECM. ScRNA-seq revealed a large number of CD8+ T cells infiltrating into the brainstem in ECM mice. Brain-infiltrated CD8+ T cells were highly activated according to scRNA-seq, immunofluorescence, and flow cytometry assays. Further analysis found a subset of ki-67+ CD8+ T cells that have a higher transcriptional level of genes related to T cell function, activation, and proliferation, suggesting that they were exposed to specific antigens presented by brain parenchyma cells. Brain-infiltrated CD8+ T cells were the only prominent source of IFN-γ in this single-cell analysis. Astrocytes, which have a high interferon response, act as cross-presenting cells to recruit and re-activate brain-infiltrated CD8+ T cells. We also found that brain-infiltrated CD8+ T cells were highly expressed immune checkpoint molecule PD-1, while parenchyma cells showed up-regulation of PD-L1 after infection. CONCLUSIONS These findings reveal a novel interaction between brain-infiltrated CD8+ T cells and parenchyma cells in the ECM brainstem, suggesting that the PD-1/PD-L1 signal pathway is a promising adjunctive therapeutic strategy for ECM targeting over-activated CD8+ T cells.
Collapse
Affiliation(s)
- Jun Wang
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| | - Qinghao Zhu
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| | - Yan Shen
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| | - Jiao Liang
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| | - Yi Wang
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| | - Yuxiao Huang
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| | - Guodong Tong
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
- College of Life SciencesNorthwest UniversityXi'anChina
| | - Xu Wang
- School of Basic Medical SciencesFourth Military Medical UniversityXi'anChina
| | - Ningning Zhang
- School of Basic Medical SciencesFourth Military Medical UniversityXi'anChina
| | - Kangjie Yu
- Department of PathologyAir Force Hospital of Eastern TheaterNanjingChina
| | - Yinghui Li
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| | - Ya Zhao
- Department of Medical Microbiology and ParasitologyFourth Military Medical UniversityXi'anChina
| |
Collapse
|
3
|
Fain CE, Zheng J, Jin F, Ayasoufi K, Wu Y, Lilley MT, Dropik AR, Wolf DM, Rodriguez RC, Aibaidula A, Tritz ZP, Bouchal SM, Pewe LL, Urban SL, Chen Y, Chang SY, Hansen MJ, Kachergus JM, Shi J, Thompson EA, Jensen HE, Harty JT, Parney IF, Sun J, Wu LJ, Johnson AJ. Discrete class I molecules on brain endothelium differentially regulate neuropathology in experimental cerebral malaria. Brain 2024; 147:566-589. [PMID: 37776513 PMCID: PMC11734323 DOI: 10.1093/brain/awad319] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/15/2023] [Accepted: 08/31/2023] [Indexed: 10/02/2023] Open
Abstract
Cerebral malaria is the deadliest complication that can arise from Plasmodium infection. CD8 T-cell engagement of brain vasculature is a putative mechanism of neuropathology in cerebral malaria. To define contributions of brain endothelial cell major histocompatibility complex (MHC) class I antigen-presentation to CD8 T cells in establishing cerebral malaria pathology, we developed novel H-2Kb LoxP and H-2Db LoxP mice crossed with Cdh5-Cre mice to achieve targeted deletion of discrete class I molecules, specifically from brain endothelium. This strategy allowed us to avoid off-target effects on iron homeostasis and class I-like molecules, which are known to perturb Plasmodium infection. This is the first endothelial-specific ablation of individual class-I molecules enabling us to interrogate these molecular interactions. In these studies, we interrogated human and mouse transcriptomics data to compare antigen presentation capacity during cerebral malaria. Using the Plasmodium berghei ANKA model of experimental cerebral malaria (ECM), we observed that H-2Kb and H-2Db class I molecules regulate distinct patterns of disease onset, CD8 T-cell infiltration, targeted cell death and regional blood-brain barrier disruption. Strikingly, ablation of either molecule from brain endothelial cells resulted in reduced CD8 T-cell activation, attenuated T-cell interaction with brain vasculature, lessened targeted cell death, preserved blood-brain barrier integrity and prevention of ECM and the death of the animal. We were able to show that these events were brain-specific through the use of parabiosis and created the novel technique of dual small animal MRI to simultaneously scan conjoined parabionts during infection. These data demonstrate that interactions of CD8 T cells with discrete MHC class I molecules on brain endothelium differentially regulate development of ECM neuropathology. Therefore, targeting MHC class I interactions therapeutically may hold potential for treatment of cases of severe malaria.
Collapse
Affiliation(s)
- Cori E Fain
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Jiaying Zheng
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905USA
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | | | - Yue Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Meredith T Lilley
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Abigail R Dropik
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Delaney M Wolf
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | | | - Abudumijiti Aibaidula
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905USA
| | - Zachariah P Tritz
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Samantha M Bouchal
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Lecia L Pewe
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Stina L Urban
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Yin Chen
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Su-Youne Chang
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905USA
| | | | | | - Ji Shi
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224USA
| | - Hadley E Jensen
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | - John T Harty
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905USA
| | - Jie Sun
- Department of Medicine, University of Virginia, Charlottesville, VA 22903USA
| | - Long-Jun Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905USA
| |
Collapse
|
4
|
Liang R, Rao H, Pang Q, Xu R, Jiao Z, Lin L, Li L, Zhong L, Zhang Y, Guo Y, Xiao N, Liu S, Chen XF, Su XZ, Li J. Human ApoE2 protects mice against Plasmodium berghei ANKA experimental cerebral malaria. mBio 2023; 14:e0234623. [PMID: 37874152 PMCID: PMC10746236 DOI: 10.1128/mbio.02346-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 10/25/2023] Open
Abstract
IMPORTANCE Cerebral malaria (CM) is the deadliest complication of malaria infection with an estimated 15%-25% mortality. Even with timely and effective treatment with antimalarial drugs such as quinine and artemisinin derivatives, survivors of CM may suffer long-term cognitive and neurological impairment. Here, we show that human apolipoprotein E variant 2 (hApoE2) protects mice from experimental CM (ECM) via suppression of CD8+ T cell activation and infiltration to the brain, enhanced cholesterol metabolism, and increased IFN-γ production, leading to reduced endothelial cell apoptosis, BBB disruption, and ECM symptoms. Our results suggest that hApoE can be an important factor for risk assessment and treatment of CM in humans.
Collapse
Affiliation(s)
- Rui Liang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Hengjun Rao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qin Pang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Ruixue Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhiwei Jiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lirong Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Li Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yixin Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yazhen Guo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Nengming Xiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Shengfa Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, China
| | - Xin-zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jian Li
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
5
|
Cimperman CK, Pena M, Gokcek SM, Theall BP, Patel MV, Sharma A, Qi C, Sturdevant D, Miller LH, Collins PL, Pierce SK, Akkaya M. Cerebral Malaria Is Regulated by Host-Mediated Changes in Plasmodium Gene Expression. mBio 2023; 14:e0339122. [PMID: 36852995 PMCID: PMC10127683 DOI: 10.1128/mbio.03391-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 03/01/2023] Open
Abstract
Cerebral malaria (CM), the deadliest complication of Plasmodium infection, is a complex and unpredictable disease. However, our understanding of the host and parasite factors that cause CM is limited. Using a mouse model of CM, experimental CM (ECM), we performed a three-way comparison between ECM-susceptible C57BL/6 mice infected with ECM-causing Plasmodium ANKA parasites [ANKA(C57BL/6)], ECM-resistant BALB/c mice infected with Plasmodium ANKA [ANKA(BALB/c)], and C57BL/6 mice infected with Plasmodium NK65 that does not cause ECM [NK65(C57BL/6)]. All ANKA(C57BL/6) mice developed CM. In contrast, in ANKA(BALB/c) and NK65(C57BL/6), infections do not result in CM and proceed similarly in terms of parasite growth, disease course, and host immune response. However, parasite gene expression in ANKA(BALB/c) was remarkably different than that in ANKA(C57BL/6) but similar to the gene expression in NK65(C57BL/6). Thus, Plasmodium ANKA has an ECM-specific gene expression profile that is activated only in susceptible hosts, providing evidence that the host has a critical influence on the outcome of infection. IMPORTANCE Hundreds of thousands of lives are lost each year due to the brain damage caused by malaria disease. The overwhelming majority of these deaths occur in young children living in sub-Saharan Africa. Thus far, there are no vaccines against this deadly disease, and we still do not know why fatal brain damage occurs in some children while others have milder, self-limiting disease progression. Our research provides an important clue to this problem. Here, we showed that the genetic background of the host has an important role in determining the course and the outcome of the disease. Our research also identified parasite molecules that can potentially be targeted in vaccination and therapy approaches.
Collapse
Affiliation(s)
- Clare K. Cimperman
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Mirna Pena
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Sohret M. Gokcek
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Brandon P. Theall
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Meha V. Patel
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Anisha Sharma
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - ChenFeng Qi
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Daniel Sturdevant
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, USA
| | - Louis H. Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Patrick L. Collins
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Susan K. Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Munir Akkaya
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
6
|
Halder SK, Milner R. Exaggerated hypoxic vascular breakdown in aged brain due to reduced microglial vasculo-protection. Aging Cell 2022; 21:e13720. [PMID: 36130175 PMCID: PMC9649604 DOI: 10.1111/acel.13720] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/04/2022] [Accepted: 08/31/2022] [Indexed: 01/25/2023] Open
Abstract
In a recent study of young mice, we showed that chronic mild hypoxia (CMH, 8% O2 ) triggers transient blood-brain barrier (BBB) disruption, and that microglia play an important vasculo-protective function in maintaining BBB integrity. As hypoxia is a common component of many age-related diseases, here we extended these studies to aged mice and found that hypoxia-induced vascular leak was greatly amplified (5-fold to 10-fold) in aged mice, being particularly high in the olfactory bulb and midbrain. While aged mice showed no obvious difference in the early stages of hypoxic angiogenic remodeling, the compensatory increase in vascularity and vessel maturation was significantly delayed. Compared with young brain, microglia in the normoxic aged brain were markedly activated, and this was further increased under hypoxic conditions, but paradoxically, this correlated with reduced vasculo-protection. Microglial depletion studies showed that microglial still play an important vasculo-protective role in aged brain, but interestingly, partial attenuation of microglial activation with minocycline resulted in fewer vascular leaks and reduced loss of endothelial tight junction proteins. Taken together, these findings suggest that increased BBB disruption in hypoxic aged mice can be explained both by a delayed vascular remodeling response and reduced microglial vasculo-protection. Importantly, they show that overly activated microglia in the aged brain are less effective at maintaining vascular integrity, though this can be improved by reducing microglial activation with minocycline, suggesting therapeutic potential for enhancing BBB integrity in the hypoxia-predisposed elderly population.
Collapse
Affiliation(s)
- Sebok K. Halder
- San Diego Biomedical Research InstituteSan DiegoCaliforniaUSA
| | - Richard Milner
- San Diego Biomedical Research InstituteSan DiegoCaliforniaUSA
| |
Collapse
|
7
|
Tian Y, Zheng Z, Wang X, Liu S, Gu L, Mu J, Zheng X, Li Y, Shen S. Establishment and evaluation of glucose-modified nanocomposite liposomes for the treatment of cerebral malaria. J Nanobiotechnology 2022; 20:318. [PMID: 35794597 PMCID: PMC9258070 DOI: 10.1186/s12951-022-01493-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 06/03/2022] [Indexed: 11/10/2022] Open
Abstract
Cerebral malaria (CM) is a life-threatening neurological complication caused by Plasmodium falciparum. About 627,000 patients died of malaria in 2020. Currently, artemisinin and its derivatives are the front-line drugs used for the treatment of cerebral malaria. However, they cannot target the brain, which decreases their effectiveness. Therefore, increasing their ability to target the brain by the nano-delivery system with brain-targeted materials is of great significance for enhancing the effects of antimalarials and reducing CM mortality. This study used glucose transporter 1 (GLUT1) on the blood-brain barrier as a target for a synthesized cholesterol-undecanoic acid-glucose conjugate. The molecular dynamics simulation found that the structural fragment of glucose in the conjugate faced the outside the phospholipid bilayers, which was conducive to the recognition of brain-targeted liposomes by GLUT1. The fluorescence intensity of the brain-targeted liposomes (na-ATS/TMP@lipoBX) in the mouse brain was significantly higher than that of the non-targeted liposomes (na-ATS/TMP@lipo) in vivo (P < 0.001) after intranasal administration. The infection and recurrence rate of the mice receiving na-ATS/TMP@lipoBX treatment were significantly decreased, which had more advantages than those of other administration groups. The analysis of pharmacokinetic data showed that na-ATS/TMP@lipoBX could enter the brain in both systemic circulation and nasal-brain pathway to treat malaria. Taken together, these results in this study provide a new approach to the treatment of cerebral malaria.
Collapse
Affiliation(s)
- Ya Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
- The Hospital of Nanbu County, Sichuan, People's Republic of China
| | - Zhongyuan Zheng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Xi Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Shuzhi Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Liwei Gu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Jing Mu
- Chinese Traditional Medicine Resource Center, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China
| | - Xiaojun Zheng
- Pharmacy Department of the first hospital of Shanxi Medical University, Shanxi, 10114, People's Republic of China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China.
| | - Shuo Shen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, People's Republic of China.
| |
Collapse
|
8
|
Wai CH, Jin J, Cyrklaff M, Genoud C, Funaya C, Sattler J, Maceski A, Meier S, Heiland S, Lanzer M, Frischknecht F, Kuhle J, Bendszus M, Hoffmann A. Neurofilament light chain plasma levels are associated with area of brain damage in experimental cerebral malaria. Sci Rep 2022; 12:10726. [PMID: 35750882 PMCID: PMC9232608 DOI: 10.1038/s41598-022-14291-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 06/03/2022] [Indexed: 11/23/2022] Open
Abstract
Neurofilament light chain (NfL), released during central nervous injury, has evolved as a powerful serum marker of disease severity in many neurological disorders, including infectious diseases. So far NfL has not been assessed in cerebral malaria in human or its rodent model experimental cerebral malaria (ECM), a disease that can lead to fatal brain edema or reversible brain edema. In this study we assessed if NfL serum levels can also grade disease severity in an ECM mouse model with reversible (n = 11) and irreversible edema (n = 10). Blood–brain-barrier disruption and brain volume were determined by magnetic resonance imaging. Neurofilament density volume as well as structural integrity were examined by electron microscopy in regions of most severe brain damage (olfactory bulb (OB), cortex and brainstem). NfL plasma levels in mice with irreversible edema (317.0 ± 45.01 pg/ml) or reversible edema (528.3 ± 125.4 pg/ml) were significantly increased compared to controls (103.4 ± 25.78 pg/ml) by three to five fold, but did not differ significantly in mice with reversible or irreversible edema. In both reversible and irreversible edema, the brain region most affected was the OB with highest level of blood–brain-barrier disruption and most pronounced decrease in neurofilament density volume, which correlated with NfL plasma levels (r = − 0.68, p = 0.045). In cortical and brainstem regions neurofilament density was only decreased in mice with irreversible edema and strongest in the brainstem. In reversible edema NfL plasma levels, MRI findings and neurofilament volume density normalized at 3 months’ follow-up. In conclusion, NfL plasma levels are elevated during ECM confirming brain damage. However, NfL plasma levels fail short on reliably indicating on the final outcomes in the acute disease stage that could be either fatal or reversible. Increased levels of plasma NfL during the acute disease stage are thus likely driven by the anatomical location of brain damage, the olfactory bulb, a region that serves as cerebral draining pathway into the nasal lymphatics.
Collapse
Affiliation(s)
- Chi Ho Wai
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Jessica Jin
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Marek Cyrklaff
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Christel Genoud
- Electron Microscopy Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Charlotta Funaya
- Electron Microscopy Core Facility, Heidelberg University, Heidelberg, Germany
| | - Julia Sattler
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Aleksandra Maceski
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stephanie Meier
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Lanzer
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Friedrich Frischknecht
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Angelika Hoffmann
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany. .,Department of Neuroradiology, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Freiburgstrasse, 3010, Bern, Switzerland.
| |
Collapse
|
9
|
Rodriguez-Muñoz D, Sánchez Á, Pérez-Benavente S, Contreras-Jurado C, Montero-Pedrazuela A, Toledo-Castillo M, Gutiérrez-Hernández M, Rodrigues-Díez R, Folgueira C, Briones AM, Sabio G, Monedero-Cobeta I, Chávez-Coira I, Castejón D, Fernández-Valle E, Regadera J, Bautista JM, Aranda A, Alemany S. Hypothyroidism confers tolerance to cerebral malaria. SCIENCE ADVANCES 2022; 8:eabj7110. [PMID: 35385300 PMCID: PMC8985923 DOI: 10.1126/sciadv.abj7110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
The modulation of the host's metabolism to protect tissue from damage induces tolerance to infections increasing survival. Here, we examined the role of the thyroid hormones, key metabolic regulators, in the outcome of malaria. Hypothyroidism confers protection to experimental cerebral malaria by a disease tolerance mechanism. Hypothyroid mice display increased survival after infection with Plasmodium berghei ANKA, diminishing intracranial pressure and brain damage, without altering pathogen burden, blood-brain barrier disruption, or immune cell infiltration. This protection is reversed by treatment with a Sirtuin 1 inhibitor, while treatment of euthyroid mice with a Sirtuin 1 activator induces tolerance and reduces intracranial pressure and lethality. This indicates that thyroid hormones and Sirtuin 1 are previously unknown targets for cerebral malaria treatment, a major killer of children in endemic malaria areas.
Collapse
Affiliation(s)
- Diego Rodriguez-Muñoz
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ángela Sánchez
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Pérez-Benavente
- Department of Biochemistry and Molecular Biology, Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Constanza Contreras-Jurado
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Departamento de Bioquímica, Facultad de Medicina, Universidad Alfonso X El Sabio, Villanueva de la Cañada, Madrid, Spain
| | - Ana Montero-Pedrazuela
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Toledo-Castillo
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Gutiérrez-Hernández
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - Raquel Rodrigues-Díez
- Departament of Pharmacology, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| | - Cintia Folgueira
- Departament of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares, ISCIII, 28029 Madrid, Spain
| | - Ana M. Briones
- Departament of Pharmacology, Instituto de Investigación Hospital La Paz, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII, Madrid, Spain
| | - Guadalupe Sabio
- Departament of Myocardial Pathophysiology, Centro Nacional de Investigaciones Cardiovasculares, ISCIII, 28029 Madrid, Spain
| | | | - Irene Chávez-Coira
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
| | - David Castejón
- MNR Unit (CAI de Bioimagen), Universidad Complutense de Madrid, Madrid, Spain
| | | | - Javier Regadera
- Department of Anatomy, Histology and Neuroscience, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology, Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Aranda
- Department of Endocrine and Nervous System Pathophysiology, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Biomedicine Unit (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCIII, Madrid, Spain
- Corresponding author. (A.A.); (S.A.)
| | - Susana Alemany
- Departament of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
- Biomedicine Unit (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
- Corresponding author. (A.A.); (S.A.)
| |
Collapse
|
10
|
Wang S, Ding B, Cui M, Yan W, Xia Q, Meng D, Shen S, Xie S, Jin H, Zhang X. Fanconi Anemia Pathway Genes Advance Cervical Cancer via Immune Regulation and Cell Adhesion. Front Cell Dev Biol 2021; 9:734794. [PMID: 34869316 PMCID: PMC8634638 DOI: 10.3389/fcell.2021.734794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/22/2021] [Indexed: 12/19/2022] Open
Abstract
Fanconi anemia (FA) pathway is a typical and multienzyme-regulated DNA damage repairer that influences the occurrence and development of disease including cancers. Few comprehensive analyses were reported about the role of FA-related genes (FARGs) and their prognostic values in cancers. In this study, a comprehensive pan-cancer analysis on 79 FARGs was performed. According to the correlation analyses between HPV integration sites and FARGs, we found that FARGs played specific and critical roles in HPV-related cancers, especially in cervical cancer (CC). Based on this, a FARGs-associated prognostic risk score (FPS) model was constructed, and subsequently a nomogram model containing the FPS was developed with a good accuracy for CC overall survival (OS) and recurrence-free survival (RFS) outcome prediction. We also used the similar expression pattern of FARGs by consensus clustering analysis to separate the patients into three subgroups that exhibited significant differential OS but not RFS. Moreover, differential expressed genes (DEGs) between the two risk groups or three clusters were identified and immune pathways as well as cell adhesion processes were determined by functional enrichment analysis. Results indicated that FARGs might promote occurrence and development of CC by regulating the immune cells' infiltration and cell adhesion. In addition, through the machine learning models containing decision tree, random forest, naïve bayes, and support vector machine models, screening of important variables on CC prognosis, we finally determined that ZBTB32 and CENPS were the main elements affecting CC OS, while PALB2 and BRCA2 were for RFS. Kaplan-Meier analysis revealed that bivariate prediction of CC outcome was reliable. Our study systematically analyzed the prognostic prediction values of FARGs and demonstrated their potential mechanism in CC aggressiveness. Results provided perspective in FA pathway-associated modification and theoretical basis for CC clinical treatments.
Collapse
Affiliation(s)
- Shizhi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Bo Ding
- Department of Gynecology and Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Mengjing Cui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Wenjing Yan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Qianqian Xia
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Dan Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Siyuan Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Shuqian Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Hua Jin
- Clinical Laboratory, Affiliated Tumor Hospital of Nantong University (Nantong Tumor Hospital), Nantong, China
| | - Xing Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China.,Department of Gynecology and Obstetrics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Clinical Laboratory, Affiliated Tumor Hospital of Nantong University (Nantong Tumor Hospital), Nantong, China
| |
Collapse
|
11
|
Matsuo-Dapaah J, Lee MSJ, Ishii KJ, Tainaka K, Coban C. Using a new three-dimensional CUBIC tissue-clearing method to examine the brain during experimental cerebral malaria. Int Immunol 2021; 33:587-594. [PMID: 34455438 DOI: 10.1093/intimm/dxab060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/28/2021] [Indexed: 11/13/2022] Open
Abstract
Cerebral malaria (CM) is a life-threatening complication of the malaria disease caused by Plasmodium falciparum infection and is responsible for the death of half a million people annually. The molecular pathogenesis underlying CM in humans is not completely understood, although sequestration of infected erythrocytes in cerebral microvessels is thought to play a major role. In contrast, experimental cerebral malaria (ECM) models in mice have been thought to be distinct from human CM, and are mainly caused by inflammatory mediators. Here, to understand the spatial distribution and the potential sequestration of parasites in the whole-brain microvessels during a mouse model of ECM, we utilized the new tissue-clearing method CUBIC (Clear, Unobstructed, Brain/Body Imaging Cocktails and Computational analysis) with light-sheet fluorescent microscopy (LSFM), and reconstructed images in three dimensions (3D). We demonstrated significantly greater accumulation of Plasmodium berghei ANKA (PbANKA) parasites in the olfactory bulb (OB) of mice, compared with the other parts of the brain, including the cerebral cortex, cerebellum and brainstem. Furthermore, we show that PbANKA parasites preferentially accumulate in the brainstem when the OB is surgically removed. This study therefore not only highlights a successful application of CUBIC tissue-clearing technology to visualize the whole brain and its microvessels during ECM, but it also shows CUBIC's future potential for visualizing pathological events in the whole ECM brain at the cellular level, an achievement that would greatly advance our understanding of human cerebral malaria.
Collapse
Affiliation(s)
- Julia Matsuo-Dapaah
- Division of Malaria Immunology, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Michelle Sue Jann Lee
- Division of Malaria Immunology, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan
| | - Ken J Ishii
- Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,International Vaccine Design Center, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Center for Bioresources, Brain Research Institute, Niigata University, Niigata, Japan.,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Cevayir Coban
- Division of Malaria Immunology, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,International Vaccine Design Center, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| |
Collapse
|
12
|
Scheunemann JF, Reichwald JJ, Korir PJ, Kuehlwein JM, Jenster LM, Hammerschmidt-Kamper C, Lewis MD, Klocke K, Borsche M, Schwendt KE, Soun C, Thiebes S, Limmer A, Engel DR, Mueller AK, Hoerauf A, Hübner MP, Schumak B. Eosinophils Suppress the Migration of T Cells Into the Brain of Plasmodium berghei-Infected Ifnar1-/- Mice and Protect Them From Experimental Cerebral Malaria. Front Immunol 2021; 12:711876. [PMID: 34659202 PMCID: PMC8514736 DOI: 10.3389/fimmu.2021.711876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
Cerebral malaria is a potentially lethal disease, which is caused by excessive inflammatory responses to Plasmodium parasites. Here we use a newly developed transgenic Plasmodium berghei ANKA (PbAAma1OVA) parasite that can be used to study parasite-specific T cell responses. Our present study demonstrates that Ifnar1-/- mice, which lack type I interferon receptor-dependent signaling, are protected from experimental cerebral malaria (ECM) when infected with this novel parasite. Although CD8+ T cell responses generated in the spleen are essential for the development of ECM, we measured comparable parasite-specific cytotoxic T cell responses in ECM-protected Ifnar1-/- mice and wild type mice suffering from ECM. Importantly, CD8+ T cells were increased in the spleens of ECM-protected Ifnar1-/- mice and the blood-brain-barrier remained intact. This was associated with elevated splenic levels of CCL5, a T cell and eosinophil chemotactic chemokine, which was mainly produced by eosinophils, and an increase in eosinophil numbers. Depletion of eosinophils enhanced CD8+ T cell infiltration into the brain and increased ECM induction in PbAAma1OVA-infected Ifnar1-/- mice. However, eosinophil-depletion did not reduce the CD8+ T cell population in the spleen or reduce splenic CCL5 concentrations. Our study demonstrates that eosinophils impact CD8+ T cell migration and proliferation during PbAAma1OVA-infection in Ifnar1-/- mice and thereby are contributing to the protection from ECM.
Collapse
Affiliation(s)
- Johanna F Scheunemann
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Julia J Reichwald
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Patricia Jebett Korir
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Janina M Kuehlwein
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Lea-Marie Jenster
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | | | - Matthew D Lewis
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Katrin Klocke
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Max Borsche
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Kim E Schwendt
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Camille Soun
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Stephanie Thiebes
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Andreas Limmer
- Clinic for Anesthesiology and Intensive Care, University Hospital Essen, Essen, Germany
| | - Daniel R Engel
- Institute for Experimental Immunology and Imaging, University Hospital Essen, Essen, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre for Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Marc P Hübner
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Beatrix Schumak
- Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
13
|
Zheng Z, Liu H, Wang X, Zhang Y, Qu S, Yang Y, Deng S, Chen L, Zhu X, Li Y. Artesunate and Tetramethylpyrazine Exert Effects on Experimental Cerebral Malaria in a Mechanism of Protein S-Nitrosylation. ACS Infect Dis 2021; 7:2836-2849. [PMID: 34254783 DOI: 10.1021/acsinfecdis.1c00085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cerebral malaria (CM) is caused by Plasmodium falciparum, resulting in severe sequelae; one of its pathogenic factors is the low bioavailability of nitric oxide (NO). Our previous study suggested that the combination of artesunate (AS) and tetramethylpyrazine (TMP) exerts an adjuvant therapeutic effect on the symptoms of experimental CM (ECM) and that NO regulation plays an important role. In the present study, we further verified the effects of AS+TMP on cerebral blood flow (CBF) and detected NO-related indicators. We focused on the role of NO through S-nitrosoproteome based on previous proteomics data and explored the mechanism of AS+TMP for improving pathological ECM symptoms. We observed that AS+TMP reduces adhesion, increases CBF, and regulates NO synthase (NOS) activity, thereby regulating the level of S-nitrosothiols, such as metabolism-related or neuro-associated receptors, for improving ECM symptoms. These results demonstrated that AS+TMP could be an effective strategy in adjuvant therapy of CM.
Collapse
Affiliation(s)
- Zhongyuan Zheng
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Hui Liu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xi Wang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yu Zhang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shuiqing Qu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuanmin Yang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shuoqiu Deng
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Lina Chen
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaoxin Zhu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yujie Li
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
14
|
Ghazanfari N, Gregory JL, Devi S, Fernandez-Ruiz D, Beattie L, Mueller SN, Heath WR. CD8 + and CD4 + T Cells Infiltrate into the Brain during Plasmodium berghei ANKA Infection and Form Long-Term Resident Memory. THE JOURNAL OF IMMUNOLOGY 2021; 207:1578-1590. [PMID: 34400523 DOI: 10.4049/jimmunol.2000773] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/18/2021] [Indexed: 12/31/2022]
Abstract
In the Plasmodium berghei ANKA mouse model of malaria, accumulation of CD8+ T cells and infected RBCs in the brain promotes the development of experimental cerebral malaria (ECM). In this study, we used malaria-specific transgenic CD4+ and CD8+ T cells to track evolution of T cell immunity during the acute and memory phases of P. berghei ANKA infection. Using a combination of techniques, including intravital multiphoton and confocal microscopy and flow cytometric analysis, we showed that, shortly before onset of ECM, both CD4+ and CD8+ T cell populations exit the spleen and begin infiltrating the brain blood vessels. Although dominated by CD8+ T cells, a proportion of both T cell subsets enter the brain parenchyma, where they are largely associated with blood vessels. Intravital imaging shows these cells moving freely within the brain parenchyma. Near the onset of ECM, leakage of RBCs into areas of the brain can be seen, implicating severe damage. If mice are cured before ECM onset, brain infiltration by T cells still occurs, but ECM is prevented, allowing development of long-term resident memory T cell populations within the brain. This study shows that infiltration of malaria-specific T cells into the brain parenchyma is associated with cerebral immunopathology and the formation of brain-resident memory T cells. The consequences of these resident memory populations is unclear but raises concerns about pathology upon secondary infection.
Collapse
Affiliation(s)
- Nazanin Ghazanfari
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Julia L Gregory
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Sapna Devi
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - William R Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and .,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
15
|
de Sousa LP, Ribeiro-Gomes FL, de Almeida RF, Souza TME, Werneck GL, Souza DO, Daniel-Ribeiro CT. Immune system challenge improves recognition memory and reverses malaria-induced cognitive impairment in mice. Sci Rep 2021; 11:14857. [PMID: 34290279 PMCID: PMC8295320 DOI: 10.1038/s41598-021-94167-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/05/2021] [Indexed: 11/09/2022] Open
Abstract
The immune system plays a role in the maintenance of healthy neurocognitive function. Different patterns of immune response triggered by distinct stimuli may affect nervous functions through regulatory or deregulatory signals, depending on the properties of the exogenous immunogens. Here, we investigate the effect of immune stimulation on cognitive-behavioural parameters in healthy mice and its impact on cognitive sequelae resulting from non-severe experimental malaria. We show that immune modulation induced by a specific combination of immune stimuli that induce a type 2 immune response can enhance long-term recognition memory in healthy adult mice subjected to novel object recognition task (NORT) and reverse a lack of recognition ability in NORT and anxiety-like behaviour in a light/dark task that result from a single episode of mild Plasmodium berghei ANKA malaria. Our findings suggest a potential use of immunogens for boosting and recovering recognition memory that may be impaired by chronic and infectious diseases and by the effects of ageing.
Collapse
Affiliation(s)
- Luciana Pereira de Sousa
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil
| | - Flávia Lima Ribeiro-Gomes
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil
| | - Roberto Farina de Almeida
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
- Programa de Pós-Graduação em Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade Federal de Ouro Preto, Minas Gerais, Brazil
| | - Tadeu Mello E Souza
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilherme Loureiro Werneck
- Departamento de Epidemiologia of Instituto de Medicina Social, Universidade do Estado do Rio de Janeiro and Instituto de Estudos de Saúde Coletiva da Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Cláudio Tadeu Daniel-Ribeiro
- Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz & Centro de Pesquisa, Diagnóstico e Treinamento em Malária (CPD-Mal) of Fundação Oswaldo Cruz (Fiocruz) and of Secretaria de Vigilância em Saúde (SVS), Ministério da Saúde, Fiocruz. Av. Brasil 4365, Manguinhos, Rio de Janeiro, RJ, CEP 2104-360, Brazil.
| |
Collapse
|
16
|
Lima MN, Freitas RJRX, Passos BABR, Darze AMG, Castro-Faria-Neto HC, Maron-Gutierrez T. Neurovascular Interactions in Malaria. Neuroimmunomodulation 2021; 28:108-117. [PMID: 33951667 DOI: 10.1159/000515557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/23/2021] [Indexed: 11/19/2022] Open
Abstract
Malaria is caused by Plasmodium infection and remains a serious public health problem worldwide, despite control efforts. Malaria can progress to severe forms, affecting multiple organs, including the brain causing cerebral malaria (CM). CM is the most severe neurological complication of malaria, and cognitive and behavior deficits are commonly reported in surviving patients. The number of deaths from malaria has been reducing in recent years, and as a consequence, neurological sequelae have been more evident. Neurological damage in malaria might be related to the neuroinflammation, characterized by glia cell activation, neuronal apoptosis and changes in the blood-brain barrier (BBB) integrity. The neurovascular unit (NVU) is responsible for maintaining the homeostasis of the BBB. Endothelial and pericytes cells in the cerebral microvasculature and neural cells, as astrocytes, neurons, and microglia, compose the NVU. The NVU can be disturbed by parasite metabolic products, such as heme and hemozoin, or cytokines that can promote activation of endothelial and glial cells and lead to increased BBB permeability and subsequently neurodegeneration. In this review, we will approach the main changes that happen in the cells of the NVU due to neuroinflammation caused by malaria infection, and elucidate how the systemic pathophysiology is involved in the onset and progression of CM.
Collapse
Affiliation(s)
- Maiara N Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Rodrigo J R X Freitas
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Beatriz A B R Passos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Ana Maria G Darze
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Hugo C Castro-Faria-Neto
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Fiocruz, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Regulatory roles of epithelial-mesenchymal interaction (EMI) during early and androgen dependent external genitalia development. Differentiation 2019; 110:29-35. [PMID: 31590136 DOI: 10.1016/j.diff.2019.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023]
Abstract
Development of external genitalia (ExG) has been a topic of long mystery in the field of organogenesis research. Early stage male and female of mouse embryos develop a common genital tubercle (GT) in the perineum whose outgrowth extends distally from the posterior cloacal regions. Concomitant with GT outgrowth, the cloaca is divided into urogenital sinus and anorectum by urorectal septum (URS) internally. The outgrowth of the GT is associated with the formation of endodermal epithelial urethral plate (UP) attached to the ventral epidermis of the GT. Such a common developmental phase is observed until around embryonic day 15.5 (E15.5) morphologically in mouse embryogenesis. Various growth factor genes, such as Fibroblast growth factor (Fgf) and Wnt genes are expressed and function during GT formation. Since the discovery of key growth factor signals and several regulatory molecules, elucidation of their functions has been achieved utilizing mouse developmental models, conditional gene knockout mouse and in vitro culture. Analyses on the phenotypes of such mouse models have revealed that several growth factor families play fundamental roles in ExG organogenesis based on the epithelial-mesenchymal interaction (EMI). More recently, EMI between developing urethral epithelia and its bilateral mesenchyme of later stages is also reported during subsequent stage of androgen-dependent male-type urethral formation in the mouse embryo. Mafb, belonging to AP-1 family and a key androgen-responsive mesenchymal gene, is identified and starts to be expressed around E14.5 when masculinization of the urethra is initiated. Mesenchymal cell condensation and migration, which are regulated by nonmuscle myosin, are shown to be essential process for masculinization. Hence, studies on EMI at various embryonic stages are important not only for early but also for subsequent masculinization of the urethra. In this review, a dynamic mode of EMI for both early and late phases of ExG development is discussed.
Collapse
|
18
|
MRI demonstrates glutamine antagonist-mediated reversal of cerebral malaria pathology in mice. Proc Natl Acad Sci U S A 2018; 115:E12024-E12033. [PMID: 30514812 DOI: 10.1073/pnas.1812909115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The deadliest complication of Plasmodium falciparum infection is cerebral malaria (CM), with a case fatality rate of 15 to 25% in African children despite effective antimalarial chemotherapy. No adjunctive treatments are yet available for this devastating disease. We previously reported that the glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON) rescued mice from experimental CM (ECM) when administered late in the infection, a time by which mice had already suffered blood-brain barrier (BBB) dysfunction, brain swelling, and hemorrhaging. Herein, we used longitudinal MR imaging to visualize brain pathology in ECM and the impact of a new DON prodrug, JHU-083, on disease progression in mice. We demonstrate in vivo the reversal of disease markers in symptomatic, infected mice following treatment, including the resolution of edema and BBB disruption, findings usually associated with a fatal outcome in children and adults with CM. Our results support the premise that JHU-083 is a potential adjunctive treatment that could rescue children and adults from fatal CM.
Collapse
|
19
|
Ghazanfari N, Mueller SN, Heath WR. Cerebral Malaria in Mouse and Man. Front Immunol 2018; 9:2016. [PMID: 30250468 PMCID: PMC6139318 DOI: 10.3389/fimmu.2018.02016] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/15/2018] [Indexed: 12/18/2022] Open
Abstract
Cerebral malaria (CM) is an acute encephalopathy caused by the malaria parasite Plasmodium falciparum, which develops in a small minority of infected patients and is responsible for the majority of deaths in African children. Despite decades of research on CM, the pathogenic mechanisms are still relatively poorly defined. Nevertheless, many studies in recent years, using a combination of animal models, in vitro cell culture work, and human patients, provide significant insight into the pathologic mechanisms leading to CM. In this review, we summarize recent findings from mouse models and human studies on the pathogenesis of CM, understanding of which may enable development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Nazanin Ghazanfari
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,The ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,The ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| | - William R Heath
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,The ARC Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Fernandes P, Howland SW, Heiss K, Hoffmann A, Hernández-Castañeda MA, Obrová K, Frank R, Wiedemann P, Bendzus M, Rénia L, Mueller AK. A Plasmodium Cross-Stage Antigen Contributes to the Development of Experimental Cerebral Malaria. Front Immunol 2018; 9:1875. [PMID: 30154793 PMCID: PMC6102508 DOI: 10.3389/fimmu.2018.01875] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 07/30/2018] [Indexed: 01/09/2023] Open
Abstract
Cerebral malaria is a complex neurological syndrome caused by an infection with Plasmodium falciparum parasites and is exclusively attributed to a series of host–parasite interactions at the pathological blood-stage of infection. In contrast, the preceding intra-hepatic phase of replication is generally considered clinically silent and thereby excluded from playing any role in the development of neurological symptoms. In this study, however, we present an antigen PbmaLS_05 that is presented to the host immune system by both pre-erythrocytic and intra-erythrocytic stages and contributes to the development of cerebral malaria in mice. Although deletion of the endogenous PbmaLS_05 prevented the development of experimental cerebral malaria (ECM) in susceptible mice after both sporozoite and infected red blood cell (iRBC) infections, we observed significant differences in contribution of the host immune response between both modes of inoculation. Moreover, PbmaLS_05-specific CD8+ T cells contributed to the development of ECM after sporozoite but not iRBC-infection, suggesting that pre-erythrocytic antigens like PbmaLS_05 can also contribute to the development of cerebral symptoms. Our data thus highlight the importance of the natural route of infection in the study of ECM, with potential implications for vaccine and therapeutic strategies against malaria.
Collapse
Affiliation(s)
- Priyanka Fernandes
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Shanshan W Howland
- Singapore Immunology Network, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Kirsten Heiss
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany.,German Centre for Infection Research (DZIF), Heidelberg, Germany
| | - Angelika Hoffmann
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Klára Obrová
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Roland Frank
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Philipp Wiedemann
- Department of Biotechnology, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Martin Bendzus
- Singapore Immunology Network, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Laurent Rénia
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology Unit, University Hospital Heidelberg, Heidelberg, Germany.,German Centre for Infection Research (DZIF), Heidelberg, Germany
| |
Collapse
|
21
|
Burrack KS, Huggins MA, Taras E, Dougherty P, Henzler CM, Yang R, Alter S, Jeng EK, Wong HC, Felices M, Cichocki F, Miller JS, Hart GT, Johnson AJ, Jameson SC, Hamilton SE. Interleukin-15 Complex Treatment Protects Mice from Cerebral Malaria by Inducing Interleukin-10-Producing Natural Killer Cells. Immunity 2018; 48:760-772.e4. [PMID: 29625893 DOI: 10.1016/j.immuni.2018.03.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 11/27/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022]
Abstract
Cerebral malaria is a deadly complication of Plasmodium infection and involves blood brain barrier (BBB) disruption following infiltration of white blood cells. During experimental cerebral malaria (ECM), mice inoculated with Plasmodium berghei ANKA-infected red blood cells develop a fatal CM-like disease caused by CD8+ T cell-mediated pathology. We found that treatment with interleukin-15 complex (IL-15C) prevented ECM, whereas IL-2C treatment had no effect. IL-15C-expanded natural killer (NK) cells were necessary and sufficient for protection against ECM. IL-15C treatment also decreased CD8+ T cell activation in the brain and prevented BBB breakdown without influencing parasite load. IL-15C induced NK cells to express IL-10, which was required for IL-15C-mediated protection against ECM. Finally, we show that ALT-803, a modified human IL-15C, mediates similar induction of IL-10 in NK cells and protection against ECM. These data identify a regulatory role for cytokine-stimulated NK cells in the prevention of a pathogenic immune response.
Collapse
Affiliation(s)
- Kristina S Burrack
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Matthew A Huggins
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Emily Taras
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Philip Dougherty
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Christine M Henzler
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55414, USA
| | - Rendong Yang
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55414, USA
| | - Sarah Alter
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Emily K Jeng
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Hing C Wong
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Martin Felices
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Geoffrey T Hart
- Center for Immunology, Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Sara E Hamilton
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA.
| |
Collapse
|
22
|
Protection from experimental cerebral malaria with a single intravenous or subcutaneous whole-parasite immunization. Sci Rep 2018; 8:3085. [PMID: 29449638 PMCID: PMC5814423 DOI: 10.1038/s41598-018-21551-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 02/05/2018] [Indexed: 12/20/2022] Open
Abstract
Cerebral malaria is a life-threatening complication of Plasmodia infection and a major cause of child mortality in Sub-Saharan Africa. We report that protection from experimental cerebral malaria in the rodent model is obtained by a single intravenous or subcutaneous whole-parasite immunization. Whole-parasite immunization with radiation-attenuated sporozoites was equally protective as immunization with non-attenuated sporozoites under chemoprophylaxis. Both immunization regimens delayed the development of blood-stage parasites, but differences in cellular and humoral immune mechanisms were observed. Single-dose whole-parasite vaccination might serve as a relatively simple and feasible immunization approach to prevent life-threatening cerebral malaria.
Collapse
|
23
|
Abstract
Systemic inflammation mediated by Plasmodium parasites is central to malaria disease and its complications. Plasmodium parasites reside in erythrocytes and can theoretically reach all host tissues via the circulation. However, actual interactions between parasitized erythrocytes and host tissues, along with the consequent damage and pathological changes, are limited locally to specific tissue sites. Such tissue specificity of the parasite can alter the outcome of malaria disease, determining whether acute or chronic complications occur. Here, we give an overview of the recent progress that has been made in understanding tissue-specific immunopathology during Plasmodium infection. As knowledge on tissue-specific host-parasite interactions accumulates, better treatment modalities and targets may emerge for intervention in malaria disease.
Collapse
|
24
|
Lee MSJ, Coban C. Unforeseen pathologies caused by malaria. Int Immunol 2017; 30:121-129. [DOI: 10.1093/intimm/dxx076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023] Open
Affiliation(s)
- Michelle Sue Jann Lee
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Japan
| | | |
Collapse
|
25
|
Fernandez-Ruiz D, Lau LS, Ghazanfari N, Jones CM, Ng WY, Davey GM, Berthold D, Holz L, Kato Y, Enders MH, Bayarsaikhan G, Hendriks SH, Lansink LIM, Engel JA, Soon MSF, James KR, Cozijnsen A, Mollard V, Uboldi AD, Tonkin CJ, de Koning-Ward TF, Gilson PR, Kaisho T, Haque A, Crabb BS, Carbone FR, McFadden GI, Heath WR. Development of a Novel CD4 + TCR Transgenic Line That Reveals a Dominant Role for CD8 + Dendritic Cells and CD40 Signaling in the Generation of Helper and CTL Responses to Blood-Stage Malaria. THE JOURNAL OF IMMUNOLOGY 2017; 199:4165-4179. [PMID: 29084838 DOI: 10.4049/jimmunol.1700186] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Abstract
We describe an MHC class II (I-Ab)-restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human (Plasmodium falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC.
Collapse
Affiliation(s)
- Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Lei Shong Lau
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Nazanin Ghazanfari
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Claerwen M Jones
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Wei Yi Ng
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Gayle M Davey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Dorothee Berthold
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Lauren Holz
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia.,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yu Kato
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Matthias H Enders
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Ganchimeg Bayarsaikhan
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia.,Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Sanne H Hendriks
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Lianne I M Lansink
- Malaria Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Jessica A Engel
- Malaria Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Megan S F Soon
- Malaria Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Kylie R James
- Malaria Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Anton Cozijnsen
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Vanessa Mollard
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alessandro D Uboldi
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Christopher J Tonkin
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | | | - Paul R Gilson
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria 3004, Australia; and
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Ashraful Haque
- Malaria Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Brendan S Crabb
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, Victoria 3004, Australia; and
| | - Francis R Carbone
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia
| | - Geoffrey I McFadden
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - William R Heath
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria 3000, Australia; .,Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
26
|
Dende C, Meena J, Nagarajan P, Nagaraj VA, Panda AK, Padmanaban G. Nanocurcumin is superior to native curcumin in preventing degenerative changes in Experimental Cerebral Malaria. Sci Rep 2017; 7:10062. [PMID: 28855623 PMCID: PMC5577147 DOI: 10.1038/s41598-017-10672-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/11/2017] [Indexed: 01/22/2023] Open
Abstract
Curcumin has many pharmacological activities despite its poor bioavailability and in vivo stability. Here, we show that a nanoformulated curcumin (PLGA-curcumin) has better therapeutic index than native curcumin in preventing the onset of neurological symptoms and delaying the death of mice in experimental cerebral malaria. Oral PLGA-curcumin was at least as effective as native curcumin at a 15-fold lower concentration in preventing the breakdown of blood-brain barrier and inhibition of brain mRNAs for inflammatory cytokines, chemokine receptor CXCR3 and its ligand CXCL10, with an increase in the anti-inflammatory cytokine IL-10. This was also reflected in serum cytokine and chemokine levels. At equivalent concentrations, a single oral dose of PLGA-curcumin was more effective in inhibiting serum IFNγ levels and enhancing IL-10 levels than native curcumin. Even at low concentrations, PLGA-curcumin was superior to native curcumin in inhibiting the sequestration of parasitized-RBCs and CD8+ T cells in the brain. A single oral dose of 5 mg PLGA-curcumin containing 350 μg of curcumin resulted in 3–4 fold higher concentration and prolonged presence of curcumin in the brain than that obtained with 5 mg of native curcumin, indicating better bioavailability of PLGA-curcumin. PLGA-curcumin has potential as an adjunct drug to treat human cerebral malaria.
Collapse
Affiliation(s)
- Chaitanya Dende
- Department of Biochemistry, Indian Institute of Science, Bengaluru, 560012, India
| | - Jairam Meena
- National Institute of Immunology, New Delhi, 110067, India
| | | | | | | | | |
Collapse
|
27
|
Kobayashi D, Endo M, Ochi H, Hojo H, Miyasaka M, Hayasaka H. Regulation of CCR7-dependent cell migration through CCR7 homodimer formation. Sci Rep 2017; 7:8536. [PMID: 28819198 PMCID: PMC5561199 DOI: 10.1038/s41598-017-09113-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 07/24/2017] [Indexed: 01/08/2023] Open
Abstract
The chemokine receptor CCR7 contributes to various physiological and pathological processes including T cell maturation, T cell migration from the blood into secondary lymphoid tissues, and tumor cell metastasis to lymph nodes. Although a previous study suggested that the efficacy of CCR7 ligand-dependent T cell migration correlates with CCR7 homo- and heterodimer formation, the exact extent of contribution of the CCR7 dimerization remains unclear. Here, by inducing or disrupting CCR7 dimers, we demonstrated a direct contribution of CCR7 homodimerization to CCR7-dependent cell migration and signaling. Induction of stable CCR7 homodimerization resulted in enhanced CCR7-dependent cell migration and CCL19 binding, whereas induction of CXCR4/CCR7 heterodimerization did not. In contrast, dissociation of CCR7 homodimerization by a novel CCR7-derived synthetic peptide attenuated CCR7-dependent cell migration, ligand-dependent CCR7 internalization, ligand-induced actin rearrangement, and Akt and Erk signaling in CCR7-expressing cells. Our study indicates that CCR7 homodimerization critically regulates CCR7 ligand-dependent cell migration and intracellular signaling in multiple cell types.
Collapse
Affiliation(s)
- Daichi Kobayashi
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.,Laboratory of Immune Molecular Function, Faculty of Science & Engineering, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan.,WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.,Department of Pharmacology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-0012, Japan
| | - Masataka Endo
- Laboratory of Immune Molecular Function, Faculty of Science & Engineering, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Hirotaka Ochi
- Laboratory of Immune Molecular Function, Faculty of Science & Engineering, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan
| | - Hironobu Hojo
- Institute for Protein Research, Osaka University, 3-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Masayuki Miyasaka
- Interdisciplinary Program for Biomedical Sciences, Institute for Academic Initiatives, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.,MediCity Research Laboratory, University of Turku, FIN-20520, Turku, Finland.,WPI Immunology Frontier Research Center, Osaka University, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Haruko Hayasaka
- Laboratory of Immune Molecular Function, Faculty of Science & Engineering, Kindai University, 3-4-1 Kowakae, Higashiosaka, Osaka, 577-8502, Japan.
| |
Collapse
|
28
|
A distinct subpopulation of CD25 - T-follicular regulatory cells localizes in the germinal centers. Proc Natl Acad Sci U S A 2017; 114:E6400-E6409. [PMID: 28698369 DOI: 10.1073/pnas.1705551114] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T-follicular helper (Tfh) cells differentiate through a multistep process, culminating in germinal center (GC) localized GC-Tfh cells that provide support to GC-B cells. T-follicular regulatory (Tfr) cells have critical roles in the control of Tfh cells and GC formation. Although Tfh-cell differentiation is inhibited by IL-2, regulatory T (Treg) cell differentiation and survival depend on it. Here, we describe a CD25- subpopulation within both murine and human PD1+CXCR5+Foxp3+ Tfr cells. It is preferentially located in the GC and can be clearly differentiated from CD25+ non-GC-Tfr, Tfh, and effector Treg (eTreg) cells by the expression of a wide range of molecules. In comparison to CD25+ Tfr and eTreg cells, CD25- Tfr cells partially down-regulate IL-2-dependent canonical Treg features, but retain suppressive function, while simultaneously up-regulating genes associated with Tfh and GC-Tfh cells. We suggest that, similar to Tfh cells, Tfr cells follow a differentiation pathway generating a mature GC-localized subpopulation, CD25- Tfr cells.
Collapse
|
29
|
Hoffmann A, Helluy X, Fischer M, Mueller AK, Heiland S, Pham M, Bendszus M, Pfeil J. In Vivo Tracking of Edema Development and Microvascular Pathology in a Model of Experimental Cerebral Malaria Using Magnetic Resonance Imaging. J Vis Exp 2017. [PMID: 28654030 DOI: 10.3791/55334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cerebral malaria is a sign of severe malarial disease and is often a harbinger of death. While aggressive management can be life-saving, the detection of cerebral malaria can be difficult. We present an experimental mouse model of cerebral malaria that shares multiple features of the human disease, including edema and microvascular pathology. Using magnetic resonance imaging (MRI), we can detect and track the blood-brain barrier disruption, edema development, and subsequent brain swelling. We describe multiple MRI techniques that can visualize these pertinent pathological changes. Thus, we show that MRI represents a valuable tool to visualize and track pathological changes, such as edema, brain swelling, and microvascular pathology, in vivo.
Collapse
Affiliation(s)
- Angelika Hoffmann
- Department of Neuroradiology, Heidelberg University Hospital; Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital;
| | - Xavier Helluy
- Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital; NeuroImaging Centre Research, Department of Neuroscience, Ruhr-University Bochum
| | - Manuel Fischer
- Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital; German Centre for Infection Research (DZIF)
| | - Sabine Heiland
- Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital
| | - Mirko Pham
- Department of Neuroradiology, Heidelberg University Hospital; Department of Neuroradiology, University of Würzburg
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital
| | - Johannes Pfeil
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital; German Centre for Infection Research (DZIF); Center for Childhood and Adolescent Medicine, General Pediatrics, Heidelberg University Hospital
| |
Collapse
|
30
|
Lee MSJ, Maruyama K, Fujita Y, Konishi A, Lelliott PM, Itagaki S, Horii T, Lin JW, Khan SM, Kuroda E, Akira S, Ishii KJ, Coban C. Plasmodium products persist in the bone marrow and promote chronic bone loss. Sci Immunol 2017; 2:2/12/eaam8093. [PMID: 28783657 DOI: 10.1126/sciimmunol.aam8093] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/28/2017] [Indexed: 12/18/2022]
Abstract
Although malaria is a life-threatening disease with severe complications, most people develop partial immunity and suffer from mild symptoms. However, incomplete recovery from infection causes chronic illness, and little is known of the potential outcomes of this chronicity. We found that malaria causes bone loss and growth retardation as a result of chronic bone inflammation induced by Plasmodium products. Acute malaria infection severely suppresses bone homeostasis, but sustained accumulation of Plasmodium products in the bone marrow niche induces MyD88-dependent inflammatory responses in osteoclast and osteoblast precursors, leading to increased RANKL expression and overstimulation of osteoclastogenesis, favoring bone resorption. Infection with a mutant parasite with impaired hemoglobin digestion that produces little hemozoin, a major Plasmodium by-product, did not cause bone loss. Supplementation of alfacalcidol, a vitamin D3 analog, could prevent the bone loss. These results highlight the risk of bone loss in malaria-infected patients and the potential benefits of coupling bone therapy with antimalarial treatment.
Collapse
Affiliation(s)
- Michelle S J Lee
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kenta Maruyama
- Laboratory of Host Defense, IFReC, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yukiko Fujita
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Aki Konishi
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Patrick M Lelliott
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sawako Itagaki
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Toshihiro Horii
- Department of Molecular Protozoology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| | - Jing-Wen Lin
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands.,Division of Pediatric Infectious Diseases, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaboration Innovation Centre, Chengdu 610041, China
| | - Shahid M Khan
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, 2333 ZA Leiden, Netherlands
| | - Etsushi Kuroda
- Laboratory of Vaccine Science, IFReC, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, IFReC, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ken J Ishii
- Laboratory of Vaccine Science, IFReC, Osaka University, Suita, Osaka 565-0871, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Cevayir Coban
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
31
|
Platt MP, Agalliu D, Cutforth T. Hello from the Other Side: How Autoantibodies Circumvent the Blood-Brain Barrier in Autoimmune Encephalitis. Front Immunol 2017; 8:442. [PMID: 28484451 PMCID: PMC5399040 DOI: 10.3389/fimmu.2017.00442] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/30/2017] [Indexed: 12/11/2022] Open
Abstract
Antibodies against neuronal receptors and synaptic proteins are associated with autoimmune encephalitides (AE) that produce movement and psychiatric disorders. In order to exert their pathological effects on neural circuits, autoantibodies against central nervous system (CNS) targets must gain access to the brain and spinal cord by crossing the blood–brain barrier (BBB), a tightly regulated gateway formed by endothelial cells lining CNS blood vessels. To date, the pathogenic mechanisms that underlie autoantibody-triggered encephalitic syndromes are poorly understood, and how autoantibodies breach the barrier remains obscure for almost all AE syndromes. The relative importance of cellular versus humoral immune mechanisms for disease pathogenesis also remains largely unexplored. Here, we review the proposed triggers for various autoimmune encephalopathies and their animal models, as well as basic structural features of the BBB and how they differ among various CNS regions, a feature that likely underlies some regional aspects of autoimmune encephalitis pathogenesis. We then discuss the routes that antibodies and immune cells employ to enter the CNS and their implications for AE. Finally, we explore future therapeutic strategies that may either preserve or restore barrier function and thereby limit immune cell and autoantibody infiltration into the CNS. Recent mechanistic insights into CNS autoantibody entry indicate promising future directions for therapeutic intervention beyond current, short-lived therapies that eliminate circulating autoantibodies.
Collapse
Affiliation(s)
- Maryann P Platt
- Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Dritan Agalliu
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.,Department of Pharmacology, Columbia University Medical Center, New York, NY, USA.,Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| | - Tyler Cutforth
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.,Columbia Translational Neuroscience Initiative, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
32
|
Perforin Expression by CD8 T Cells Is Sufficient To Cause Fatal Brain Edema during Experimental Cerebral Malaria. Infect Immun 2017; 85:IAI.00985-16. [PMID: 28264905 DOI: 10.1128/iai.00985-16] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 02/26/2017] [Indexed: 01/11/2023] Open
Abstract
Human cerebral malaria (HCM) is a serious complication of Plasmodium falciparum infection. The most severe outcomes for patients include coma, permanent neurological deficits, and death. Recently, a large-scale magnetic resonance imaging (MRI) study in humans identified brain swelling as the most prominent predictor of fatal HCM. Therefore, in this study, we sought to define the mechanism controlling brain edema through the use of the murine experimental cerebral malaria (ECM) model. Specifically, we investigated the ability of CD8 T cells to initiate brain edema during ECM. We determined that areas of blood-brain barrier (BBB) permeability colocalized with a reduction of the cerebral endothelial cell tight-junction proteins claudin-5 and occludin. Furthermore, through small-animal MRI, we analyzed edema and vascular leakage. Using gadolinium-enhanced T1-weighted MRI, we determined that vascular permeability is not homogeneous but rather confined to specific regions of the brain. Our findings show that BBB permeability was localized within the brainstem, olfactory bulb, and lateral ventricle. Concurrently with the initiation of vascular permeability, T2-weighted MRI revealed edema and brain swelling. Importantly, ablation of the cytolytic effector molecule perforin fully protected against vascular permeability and edema. Furthermore, perforin production specifically by CD8 T cells was required to cause fatal edema during ECM. We propose that CD8 T cells initiate BBB breakdown through perforin-mediated disruption of tight junctions. In turn, leakage from the vasculature into the parenchyma causes brain swelling and edema. This results in a breakdown of homeostatic maintenance that likely contributes to ECM pathology.
Collapse
|
33
|
Swanson PA, Hart GT, Russo MV, Nayak D, Yazew T, Peña M, Khan SM, Janse CJ, Pierce SK, McGavern DB. CD8+ T Cells Induce Fatal Brainstem Pathology during Cerebral Malaria via Luminal Antigen-Specific Engagement of Brain Vasculature. PLoS Pathog 2016; 12:e1006022. [PMID: 27907215 PMCID: PMC5131904 DOI: 10.1371/journal.ppat.1006022] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/25/2016] [Indexed: 01/01/2023] Open
Abstract
Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection that results in thousands of deaths each year, mostly in African children. The in vivo mechanisms underlying this fatal condition are not entirely understood. Using the animal model of experimental cerebral malaria (ECM), we sought mechanistic insights into the pathogenesis of CM. Fatal disease was associated with alterations in tight junction proteins, vascular breakdown in the meninges / parenchyma, edema, and ultimately neuronal cell death in the brainstem, which is consistent with cerebral herniation as a cause of death. At the peak of ECM, we revealed using intravital two-photon microscopy that myelomonocytic cells and parasite-specific CD8+ T cells associated primarily with the luminal surface of CNS blood vessels. Myelomonocytic cells participated in the removal of parasitized red blood cells (pRBCs) from cerebral blood vessels, but were not required for the disease. Interestingly, the majority of disease-inducing parasite-specific CD8+ T cells interacted with the lumen of brain vascular endothelial cells (ECs), where they were observed surveying, dividing, and arresting in a cognate peptide-MHC I dependent manner. These activities were critically dependent on IFN-γ, which was responsible for activating cerebrovascular ECs to upregulate adhesion and antigen-presenting molecules. Importantly, parasite-specific CD8+ T cell interactions with cerebral vessels were impaired in chimeric mice rendered unable to present EC antigens on MHC I, and these mice were in turn resistant to fatal brainstem pathology. Moreover, anti-adhesion molecule (LFA-1 / VLA-4) therapy prevented fatal disease by rapidly displacing luminal CD8+ T cells from cerebrovascular ECs without affecting extravascular T cells. These in vivo data demonstrate that parasite-specific CD8+ T cell-induced fatal vascular breakdown and subsequent neuronal death during ECM is associated with luminal, antigen-dependent interactions with cerebrovasculature. Cerebral malaria (CM) is a severe and potentially fatal complication of malaria in humans that results in swelling and bleeding within the brain. The mechanisms that cause this fatal disease in humans are not completely understood. We studied an animal model known as experimental cerebral malaria to learn more about the factors that drive this disease process. Using a technique referred to as intravital microscopy, we captured movies of immune cells operating in the living brain as the disease developed. At the peak of disease, we observed evidence of immune cells interacting with and aggregating along blood vessels throughout the brain. These interactions were directly associated vascular leakage. This caused the brain to swell, which gave rise to an unsustainable pressure that ultimately killed neurons responsible for heart and lung function. The fatal swelling was induced by immune cells (referred to as T cells) interacting with bits of parasite presented by blood vessels in the brain. Removal of this parasite presentation protected the mice from fatal disease. We also evaluated a straightforward therapy that involved intravenous administration of antibodies that interfered with T cell sticking to blood vessels. Our movies revealed that this therapeutic approach rapidly displaced T cells from the blood vessels in the brain and prevented fatal disease.
Collapse
Affiliation(s)
- Phillip A. Swanson
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Geoffrey T. Hart
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Matthew V. Russo
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Debasis Nayak
- Center for Bioscience and Biomedical Engineering, Indian Institute of Technology Indore, Madhya Pradesh, India
| | - Takele Yazew
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Mirna Peña
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Shahid M. Khan
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Susan K. Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Dorian B. McGavern
- Viral Immunology & Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
34
|
Oxidative insult can induce malaria-protective trait of sickle and fetal erythrocytes. Nat Commun 2016; 7:13401. [PMID: 27824335 PMCID: PMC5105170 DOI: 10.1038/ncomms13401] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 09/29/2016] [Indexed: 01/06/2023] Open
Abstract
Plasmodium falciparum infections can cause severe malaria, but not every infected person develops life-threatening complications. In particular, carriers of the structural haemoglobinopathies S and C and infants are protected from severe disease. Protection is associated with impaired parasite-induced host actin reorganization, required for vesicular trafficking of parasite-encoded adhesins, and reduced cytoadherence of parasitized erythrocytes in the microvasculature. Here we show that aberrant host actin remodelling and the ensuing reduced cytoadherence result from a redox imbalance inherent to haemoglobinopathic and fetal erythrocytes. We further show that a transient oxidative insult to wild-type erythrocytes before infection with P. falciparum induces the phenotypic features associated with the protective trait of haemoglobinopathic and fetal erythrocytes. Moreover, pretreatment of mice with the pro-oxidative nutritional supplement menadione mitigate the development of experimental cerebral malaria. Our results identify redox imbalance as a causative principle of protection from severe malaria, which might inspire host-directed intervention strategies.
Collapse
|
35
|
Dalko E, Genete D, Auger F, Dovergne C, Lambert C, Herbert F, Cazenave PA, Roland J, Pied S. Heme dampens T-cell sequestration by modulating glial cell responses during rodent cerebral malaria. Brain Behav Immun 2016; 58:280-290. [PMID: 27477919 DOI: 10.1016/j.bbi.2016.07.157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/21/2016] [Accepted: 07/27/2016] [Indexed: 01/24/2023] Open
Abstract
Cerebral malaria is the deadliest complication of Plasmodium falciparum infection. Its pathophysiology is associated with a strong pro-inflammatory reaction and the activation of glial cells. Among modulators released during the infection, heme seems to play a controversial role in the pathophysiology of malaria. Herein, we first investigated the phenotype of glial cells during cerebral malaria in C57BL/6 mice infected with P. berghei ANKA. Given the fact that high levels of heme were associated with cerebral malaria, we then investigated its impact on microglial, astrocyte, and T cell responses to further clarify its contribution in the neuropathophysiology. Surprisingly, we found that administration of heme twice a day from day three of infection induced the expression of the Heme oxygenase-1 (Hmox1) gene and prevented brain damages. More specifically, heme inhibited the M1 phenotype of microglia, hampered the activation of astrocytes, and decreased the cerebral expression of Ifng, Tnfa and Ip10. Heme might that way alter the migration of pathogenic CD4 and CD8 T lymphocytes within the brain observed during cerebral malaria. Taking into account that cerebral malaria results from a complex interplay between host- and parasite-derived factors, it is possible that genetic polymorphisms of Hmox1, which could be associated with the control of systemic levels of heme during P. falciparum infection, might explain its dual role and its contribution to the resistance to cerebral malaria.
Collapse
Affiliation(s)
- Esther Dalko
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France; Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, Canada
| | - Delphine Genete
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Florent Auger
- Imaging Platform, Research Pole, Lille University Hospital, Lille, France; Université de Lille, UDSL, EA 1046 Lille, France
| | - Claire Dovergne
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Claire Lambert
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Fabien Herbert
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Pierre-André Cazenave
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Jacques Roland
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France
| | - Sylviane Pied
- Center for Infection and Immunity of Lille, CNRS UMR 8204, Université de Lille, Institut Pasteur de Lille, Lille, France.
| |
Collapse
|
36
|
Shrivastava SK, Dalko E, Delcroix-Genete D, Herbert F, Cazenave PA, Pied S. Uptake of parasite-derived vesicles by astrocytes and microglial phagocytosis of infected erythrocytes may drive neuroinflammation in cerebral malaria. Glia 2016; 65:75-92. [PMID: 27696532 DOI: 10.1002/glia.23075] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 09/12/2016] [Indexed: 01/01/2023]
Abstract
Astrocytes and microglia are activated during cerebral malaria (CM) and contribute to the production and release of several mediators during neuroinflammatory processes. Whether these changes are the consequence of a direct crosstalk between glial cells and the malarial parasite and how these cells participate in the pathogenesis of CM is not yet clear. We therefore examined the interaction of astrocytes and microglia with Plasmodium berghei ANKA-infected red blood cells using primary cell cultures derived from newborn C57BL/6 mice. We observed a dynamic transfer of vesicles from the parasite to astrocytes within minutes of contact, and the phagocytosis of infected red blood cells by microglia. Differential gene expression studies using the Affymetrix GeneChip® microarray, and quantitative PCR analyses showed the increase in expression of the set of genes belonging to the immune response network in parasite activated astrocytes and microglia. Interestingly, expression of these genes was also significantly upregulated in brains of mice dying from CM compared with uninfected mice or infected mice that did not develop the neuropathology. Accumulation of parasite-derived vesicles within astrocytes, and the phagocytosis of infected red blood cells by microglia induced a subsequent increase in interferon gamma inducible protein 10 (IP10) in both the brain and plasma of infected mice at the onset of CM, confirming a role for this molecule in CM pathogenesis. Altogether, these observations point to a possible role for glial cells in the neuropathological processes leading to CM. GLIA 2016 GLIA 2017;65:75-92.
Collapse
Affiliation(s)
- Sandeep K Shrivastava
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, F-59000, France
| | - Esther Dalko
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, F-59000, France
| | - Delphine Delcroix-Genete
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, F-59000, France
| | - Fabien Herbert
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, F-59000, France
| | - Pierre-André Cazenave
- Unité d'Immunophysiopathologie Infectieuse, CRNS URA 1961, UPMC, Institut Pasteur, Paris, France
| | - Sylviane Pied
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, F-59000, France.,Unité d'Immunophysiopathologie Infectieuse, CRNS URA 1961, UPMC, Institut Pasteur, Paris, France
| |
Collapse
|
37
|
DellaValle B, Hempel C, Staalsoe T, Johansen FF, Kurtzhals JAL. Glucagon-like peptide-1 analogue, liraglutide, in experimental cerebral malaria: implications for the role of oxidative stress in cerebral malaria. Malar J 2016; 15:427. [PMID: 27554094 PMCID: PMC4995661 DOI: 10.1186/s12936-016-1486-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/11/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cerebral malaria from Plasmodium falciparum infection is major cause of death in the tropics. The pathogenesis of the disease is complex and the contribution of reactive oxygen and nitrogen species (ROS/RNS) in the brain is incompletely understood. Insulinotropic glucagon-like peptide-1 (GLP-1) mimetics have potent neuroprotective effects in animal models of neuropathology associated with ROS/RNS dysfunction. This study investigates the effect of the GLP-1 analogue, liraglutide against the clinical outcome of experimental cerebral malaria (ECM) and Plasmodium falciparum growth. Furthermore the role of oxidative stress on ECM pathogenesis is evaluated. METHODS ECM was induced in Plasmodium berghei ANKA-infected C57Bl/6j mice. Infected Balb/c (non-cerebral malaria) and uninfected C57Bl/6j mice were included as controls. Mice were treated twice-daily with vehicle or liraglutide (200 μg/kg). ROS/RNS were quantified with in vivo imaging and further analyzed ex vivo. Brains were assayed for cAMP, activation of cAMP response element binding protein (CREB) and nitrate/nitrite. Plasmodium falciparum was cultivated in vitro with increasing doses of liraglutide and growth and metabolism were quantified. RESULTS The development and progression of ECM was not affected by liraglutide. Indeed, although ROS/RNS were increased in peripheral organs, ROS/RNS generation was not present in the brain. Interestingly, CREB was activated in the ECM brain and may protect against ROS/RNS stress. Parasite growth was not adversely affected by liraglutide in mice or in P. falciparum cultures indicating safety should not be a concern in type-II diabetics in endemic regions. CONCLUSIONS Despite the breadth of models where GLP-1 is neuroprotective, ECM was not affected by liraglutide providing important insight into the pathogenesis of ECM. Furthermore, ECM does not induce excess ROS/RNS in the brain potentially associated with activation of the CREB system.
Collapse
Affiliation(s)
- Brian DellaValle
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark. .,Department of Biomedical Sciences, Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark. .,Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Casper Hempel
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Trine Staalsoe
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Flemming Fryd Johansen
- Department of Biomedical Sciences, Biotech Research and Innovation Center, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Anders Lindholm Kurtzhals
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
38
|
Experimental Cerebral Malaria Spreads along the Rostral Migratory Stream. PLoS Pathog 2016; 12:e1005470. [PMID: 26964100 PMCID: PMC4786214 DOI: 10.1371/journal.ppat.1005470] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 02/03/2016] [Indexed: 12/25/2022] Open
Abstract
It is poorly understood how progressive brain swelling in experimental cerebral malaria (ECM) evolves in space and over time, and whether mechanisms of inflammation or microvascular sequestration/obstruction dominate the underlying pathophysiology. We therefore monitored in the Plasmodium berghei ANKA-C57BL/6 murine ECM model, disease manifestation and progression clinically, assessed by the Rapid-Murine-Coma-and-Behavioral-Scale (RMCBS), and by high-resolution in vivo MRI, including sensitive assessment of early blood-brain-barrier-disruption (BBBD), brain edema and microvascular pathology. For histological correlation HE and immunohistochemical staining for microglia and neuroblasts were obtained. Our results demonstrate that BBBD and edema initiated in the olfactory bulb (OB) and spread along the rostral-migratory-stream (RMS) to the subventricular zone of the lateral ventricles, the dorsal-migratory-stream (DMS), and finally to the external capsule (EC) and brainstem (BS). Before clinical symptoms (mean RMCBS = 18.5±1) became evident, a slight, non-significant increase of quantitative T2 and ADC values was observed in OB+RMS. With clinical manifestation (mean RMCBS = 14.2±0.4), T2 and ADC values significantly increased along the OB+RMS (p = 0.049/p = 0.01). Severe ECM (mean RMCBS = 5±2.9) was defined by further spread into more posterior and deeper brain structures until reaching the BS (significant T2 elevation in DMS+EC+BS (p = 0.034)). Quantitative automated histological analyses confirmed microglial activation in areas of BBBD and edema. Activated microglia were closely associated with the RMS and neuroblasts within the RMS were severely misaligned with respect to their physiological linear migration pattern. Microvascular pathology and ischemic brain injury occurred only secondarily, after vasogenic edema formation and were both associated less with clinical severity and the temporal course of ECM. Altogether, we identified a distinct spatiotemporal pattern of microglial activation in ECM involving primarily the OB+RMS axis, a distinct pathway utilized by neuroblasts and immune cells. Our data suggest significant crosstalk between these two cell populations to be operative in deeper brain infiltration and further imply that the manifestation and progression of cerebral malaria may depend on brain areas otherwise serving neurogenesis. Brain swelling is difficult to detect ex vivo and has recently been identified as a strong predictor of death not only in experimental cerebral malaria (ECM), but also in human cerebral malaria. As whole-brain in-vivo imaging methods have been widely underutilized in this disease model, little is known about the spatiotemporal evolution of brain swelling. To unravel this question, we monitored the evolution of ECM in vivo using high-field magnetic resonance imaging (MRI) with whole-brain coverage and have identified a distinct pattern of cerebral disease spread. Inflammatory disruption of the blood-brain-barrier and consecutive brain swelling initiates in the olfactory bulb and spreads from there along the rostral migratory stream—a neurogenic niche—deeper into the brain. When the brainstem is eventually reached, mice start to fall into a comatose state. Those findings correlate with previously published human MRI findings, which also show brain swelling of the brainstem in comatose children with cerebral malaria as well as early involvement of the striatum—recently recognized to serve neurogenesis in humans. Our study provides a novel link between neurogenic areas specifically permitting the spatiotemporal expansion of activated microglia, blood-brain-barrier disruption and consequent brain edema. Finally, the dominant role of the neurogenic axis in the transmission of inflammation may provide an explanation why children are more vulnerable to cerebral malaria.
Collapse
|
39
|
Abstract
We have recently demonstrated that brain endothelial cells cross-present parasite antigen during mouse experimental cerebral malaria (ECM). Here we describe a 2-d protocol to detect cross-presentation by isolating the brain microvessels and incubating them with a reporter cell line that expresses lacZ upon detection of the relevant peptide-major histocompatibility complex. After X-gal staining, a typical positive result consists of hundreds of blue spots, compared with fewer than 20 spots from a naive brain. The assay is generalizable to other disease contexts by using reporter cells that express appropriate specific T cell receptors. Also described is the protocol for culturing endothelial cells from brain microvessels isolated from naive mice. After 7-10 d, an in vitro cross-presentation assay can be performed by adding interferon-γ, antigen (e.g., Plasmodium berghei-infected red blood cells) and reporter cells in sequence over 3 d. This is useful for comparing different antigen forms or for probing the effects of various interventions.
Collapse
|
40
|
In vivo curative and protective potential of orally administered 5-aminolevulinic acid plus ferrous ion against malaria. Antimicrob Agents Chemother 2015; 59:6960-7. [PMID: 26324278 PMCID: PMC4604406 DOI: 10.1128/aac.01910-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 08/20/2015] [Indexed: 12/30/2022] Open
Abstract
5-Aminolevulinic acid (ALA) is a naturally occurring amino acid present in diverse organisms and a precursor of heme biosynthesis. ALA is commercially available as a component of cosmetics, dietary supplements, and pharmaceuticals for cancer diagnosis and therapy. Recent reports demonstrated that the combination of ALA and ferrous ion (Fe2+) inhibits the in vitro growth of the human malaria parasite Plasmodium falciparum. To further explore the potential application of ALA and ferrous ion as a combined antimalarial drug for treatment of human malaria, we conducted an in vivo efficacy evaluation. Female C57BL/6J mice were infected with the lethal strain of rodent malaria parasite Plasmodium yoelii 17XL and orally administered ALA plus sodium ferrous citrate (ALA/SFC) as a once-daily treatment. Parasitemia was monitored in the infected mice, and elimination of the parasites was confirmed using diagnostic PCR. Treatment of P. yoelii 17XL-infected mice with ALA/SFC provided curative efficacy in 60% of the mice treated with ALA/SFC at 600/300 mg/kg of body weight; no mice survived when treated with vehicle alone. Interestingly, the cured mice were protected from homologous rechallenge, even when reinfection was attempted more than 230 days after the initial recovery, indicating long-lasting resistance to reinfection with the same parasite. Moreover, parasite-specific antibodies against reported vaccine candidate antigens were found and persisted in the sera of the cured mice. These findings provide clear evidence that ALA/SFC is effective in an experimental animal model of malaria and may facilitate the development of a new class of antimalarial drug.
Collapse
|
41
|
Simultaneously targeting inflammatory response and parasite sequestration in brain to treat Experimental Cerebral Malaria. Sci Rep 2015; 5:12671. [PMID: 26227888 PMCID: PMC4521148 DOI: 10.1038/srep12671] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/01/2015] [Indexed: 11/08/2022] Open
Abstract
Malaria afflicts around 200 million people annually, with a mortality number close to 600,000. The mortality rate in Human Cerebral Malaria (HCM) is unacceptably high (15–20%), despite the availability of artemisinin-based therapy. An effective adjunct therapy is urgently needed. Experimental Cerebral Malaria (ECM) in mice manifests many of the neurological features of HCM. Migration of T cells and parasite-infected RBCs (pRBCs) into the brain are both necessary to precipitate the disease. We have been able to simultaneously target both these parameters of ECM. Curcumin alone was able to reverse all the parameters investigated in this study that govern inflammatory responses, CD8+ T cell and pRBC sequestration into the brain and blood brain barrier (BBB) breakdown. But the animals eventually died of anemia due to parasite build-up in blood. However, arteether-curcumin (AC) combination therapy even after the onset of symptoms provided complete cure. AC treatment is a promising therapeutic option for HCM.
Collapse
|
42
|
Activated Brain Endothelial Cells Cross-Present Malaria Antigen. PLoS Pathog 2015; 11:e1004963. [PMID: 26046849 PMCID: PMC4457820 DOI: 10.1371/journal.ppat.1004963] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/19/2015] [Indexed: 12/31/2022] Open
Abstract
In the murine model of cerebral malaria caused by P. berghei ANKA (PbA), parasite-specific CD8+ T cells directly induce pathology and have long been hypothesized to kill brain endothelial cells that have internalized PbA antigen. We previously reported that brain microvessel fragments from infected mice cross-present PbA epitopes, using reporter cells transduced with epitope-specific T cell receptors. Here, we confirm that endothelial cells are the population responsible for cross-presentation in vivo, not pericytes or microglia. PbA antigen cross-presentation by primary brain endothelial cells in vitro confers susceptibility to killing by CD8+ T cells from infected mice. IFNγ stimulation is required for brain endothelial cross-presentation in vivo and in vitro, which occurs by a proteasome- and TAP-dependent mechanism. Parasite strains that do not induce cerebral malaria were phagocytosed and cross-presented less efficiently than PbA in vitro. The main source of antigen appears to be free merozoites, which were avidly phagocytosed. A human brain endothelial cell line also phagocytosed P. falciparum merozoites. Besides being the first demonstration of cross-presentation by brain endothelial cells, our results suggest that interfering with merozoite phagocytosis or antigen processing may be effective strategies for cerebral malaria intervention.
Collapse
|
43
|
Matz JM, Kooij TWA. Towards genome-wide experimental genetics in the in vivo malaria model parasite Plasmodium berghei. Pathog Glob Health 2015; 109:46-60. [PMID: 25789828 DOI: 10.1179/2047773215y.0000000006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Plasmodium berghei was identified as a parasite of thicket rats (Grammomys dolichurus) and Anopheles dureni mosquitoes in African highland forests. Successful adaptation to a range of rodent and mosquito species established P. berghei as a malaria model parasite. The introduction of stable transfection technology, permitted classical reverse genetics strategies and thus systematic functional profiling of the gene repertoire. In the past 10 years following the publication of the P. berghei genome sequence, many new tools for experimental genetics approaches have been developed and existing ones have been improved. The infection of mice is the principal limitation towards a genome-wide repository of mutant parasite lines. In the past few years, there have been some promising and most welcome developments that allow rapid selection and isolation of recombinant parasites while simultaneously minimising animal usage. Here, we provide an overview of all the currently available tools and methods.
Collapse
|
44
|
Marijon A, Bonnot G, Fourier A, Bringer C, Lavoignat A, Gagnieu MC, Bienvenu AL, Picot S. Efficacy of intranasal administration of artesunate in experimental cerebral malaria. Malar J 2014; 13:501. [PMID: 25516091 PMCID: PMC4320559 DOI: 10.1186/1475-2875-13-501] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/12/2014] [Indexed: 11/27/2022] Open
Abstract
Background Improving management of patients suffering from cerebral malaria is needed to reduce the devastating mortality and morbidity of the disease in endemic areas. Intravenous artesunate is currently the first-line treatment, but the lack of material and skills in the field make it difficult to implement in endemic areas. Intranasal route provides a very easy and direct gateway to blood and brain to deliver medications, by-passing the brain blood barrier. Therefore, it could be helpful and suitable to administer artesunate in the context of cerebral malaria, especially in young children. In this study, intranasal administration of artesunate to rescue from cerebral malaria using a murine model was tested. Methods CBA/J mice infected with Plasmodium berghei ANKA strain received artesunate (20 mg/kg) or a placebo solution intranasally, either on day 5, 6 or 7 post-infection, during a controlled, blinded, randomized trial. Primary endpoint was mortality on day 12 post-infection. Secondary endpoints were parasitaemia and clinical stage. Pharmacokinetics data following administration were collected in blood and brains of treated mice. Local toxicity was evaluated by histopathologic examination of brain and nasal sections in blinded manner. Results Intranasal administration of artesunate dramatically reduced the mortality rate (p < 0.001), preventing death in most cases. Parasitaemia loads decreased by 88.7% (61.8-100%) within 24 hours after administration. Symptoms of cerebral malaria were prevented or reversed. Dihydroartemisinin was detected in mice blood and brain within 15 minutes of intranasal administration. No direct nasal or brain toxicity was detected. Conclusion Intranasal delivery is an efficient route to timely and efficiently administer artesunate and therefore may contribute to decreasing malaria-related mortality.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stéphane Picot
- Université Claude Bernard Lyon 1, Malaria Research Unit, SMITH, ICBMS, UMR 5246 CNRS-INSA-CPE-UCBL1, 8 avenue Rockefeller, 69373 Lyon cedex 08, France.
| |
Collapse
|
45
|
Nacer A, Movila A, Sohet F, Girgis NM, Gundra UM, Loke P, Daneman R, Frevert U. Experimental cerebral malaria pathogenesis--hemodynamics at the blood brain barrier. PLoS Pathog 2014; 10:e1004528. [PMID: 25474413 PMCID: PMC4256476 DOI: 10.1371/journal.ppat.1004528] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/17/2014] [Indexed: 12/16/2022] Open
Abstract
Cerebral malaria claims the lives of over 600,000 African children every year. To better understand the pathogenesis of this devastating disease, we compared the cellular dynamics in the cortical microvasculature between two infection models, Plasmodium berghei ANKA (PbA) infected CBA/CaJ mice, which develop experimental cerebral malaria (ECM), and P. yoelii 17XL (PyXL) infected mice, which succumb to malarial hyperparasitemia without neurological impairment. Using a combination of intravital imaging and flow cytometry, we show that significantly more CD8(+) T cells, neutrophils, and macrophages are recruited to postcapillary venules during ECM compared to hyperparasitemia. ECM correlated with ICAM-1 upregulation on macrophages, while vascular endothelia upregulated ICAM-1 during ECM and hyperparasitemia. The arrest of large numbers of leukocytes in postcapillary and larger venules caused microrheological alterations that significantly restricted the venous blood flow. Treatment with FTY720, which inhibits vascular leakage, neurological signs, and death from ECM, prevented the recruitment of a subpopulation of CD45(hi) CD8(+) T cells, ICAM-1(+) macrophages, and neutrophils to postcapillary venules. FTY720 had no effect on the ECM-associated expression of the pattern recognition receptor CD14 in postcapillary venules suggesting that endothelial activation is insufficient to cause vascular pathology. Expression of the endothelial tight junction proteins claudin-5, occludin, and ZO-1 in the cerebral cortex and cerebellum of PbA-infected mice with ECM was unaltered compared to FTY720-treated PbA-infected mice or PyXL-infected mice with hyperparasitemia. Thus, blood brain barrier opening does not involve endothelial injury and is likely reversible, consistent with the rapid recovery of many patients with CM. We conclude that the ECM-associated recruitment of large numbers of activated leukocytes, in particular CD8(+) T cells and ICAM(+) macrophages, causes a severe restriction in the venous blood efflux from the brain, which exacerbates the vasogenic edema and increases the intracranial pressure. Thus, death from ECM could potentially occur as a consequence of intracranial hypertension.
Collapse
Affiliation(s)
- Adéla Nacer
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, United States of America
| | - Alexandru Movila
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, United States of America
| | - Fabien Sohet
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
| | - Natasha M. Girgis
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, United States of America
| | - Uma Mahesh Gundra
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, United States of America
| | - P'ng Loke
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, United States of America
| | - Richard Daneman
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America
| | - Ute Frevert
- Department of Microbiology, Division of Medical Parasitology, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
46
|
Frevert U, Nacer A. Fatal cerebral malaria: a venous efflux problem. Front Cell Infect Microbiol 2014; 4:155. [PMID: 25414834 PMCID: PMC4222339 DOI: 10.3389/fcimb.2014.00155] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/13/2014] [Indexed: 12/20/2022] Open
Abstract
Most Plasmodium falciparum-infected children with cerebral malaria (CM) die from respiratory arrest, but the underlying pathology is unclear. Here we present a model in which the ultimate cause of death from CM is severe intracranial hypertension. Dynamic imaging of mice infected with P. berghei ANKA, an accepted model for experimental CM, revealed that leukocyte adhesion impairs the venous blood flow by reducing the functional lumen of postcapillary venules (PCV). The resulting increase in intracranial pressure (ICP) exacerbates cerebral edema formation, a hallmark of both murine and pediatric CM. We propose that two entirely different pathogenetic mechanisms-cytoadherence of P. falciparum-infected erythrocytes in pediatric CM and leukocyte arrest in murine CM-result in the same pathological outcome: a severe increase in ICP leading to brainstem herniation and death from respiratory arrest. The intracranial hypertension (IH) model unifies previous hypotheses, applies to human and experimental CM alike, eliminates the need to explain any selective recognition mechanism Plasmodium might use to target multiple sensitive sites in the brain, and explains how an intravascular parasite can cause so much neuronal dysfunction.
Collapse
Affiliation(s)
- Ute Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine New York, NY, USA
| | - Adéla Nacer
- Unité de Biologie des Interactions Hôte-Parasite, Département de Parasitologie et Mycologie, Institut Pasteur Paris, France
| |
Collapse
|
47
|
Targeting the olfactory bulb during experimental cerebral malaria. Trends Parasitol 2014; 30:375-6. [PMID: 24856292 DOI: 10.1016/j.pt.2014.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/07/2014] [Indexed: 11/23/2022]
Abstract
Malaria is responsible for over 500 million clinical cases and over 500000 deaths annually. Fatalities arise from a range of overlapping syndromes, such as cerebral malaria, whose pathogenesis is still incompletely understood. In a new study, Coban and colleagues provide new clues on the involvement of the olfactory bulb during experimental cerebral malaria in mice that open the way to testable hypotheses and potentially earlier intervention in humans.
Collapse
|